Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Aug 1.
Published in final edited form as: Bioorg Med Chem Lett. 2017 Jun 13;27(15):3254–3258. doi: 10.1016/j.bmcl.2017.06.033

The structural requirements of histone deacetylase inhibitors: SAHA analogs modified at the C5 position display dual HDAC6/8 selectivity

Ahmed T Negmeldin a, Kay H Pflum a,*
PMCID: PMC5747257  NIHMSID: NIHMS887586  PMID: 28648461

Abstract

Histone deacetylase (HDAC) proteins have emerged as important targets for anti-cancer drugs, with four small molecules approved for use in the clinic. Suberoylanilide hydroxamic acid (Vorinostat, SAHA) was the first FDA-approved HDAC inhibitor for cancer treatment. However, SAHA inhibits most of the eleven HDAC isoforms. To understand the structural requirements of HDAC inhibitor selectivity and develop isoform selective HDAC inhibitors, SAHA analogs modified in the linker at the C5 position were synthesized and tested for potency and selectivity. C5-modified SAHA analogs displayed dual selectivity to HDAC6 and HDAC8 over HDAC 1, 2, and 3, with only a modest reduction in potency. These findings are consistent with prior work showing that modification of the linker region of SAHA can alter isoform selectivity. The observed HDAC6/8 selectivity of C5-modified SAHA analogs provide guidance toward development of isoform selective HDAC inhibitors and more effective anti-cancer drugs.

Keywords: Histone Deacetylase, HDAC inhibitors, Vorinostat, Isoform selectivity, HDAC6/8 selective inhibitor

Graphical Abstract

graphic file with name nihms887586u1.jpg


Histone deacetylase (HDAC) proteins have a prominent regulatory role in gene transcription and cell function. HDAC proteins catalyze the removal of the acetyl group from ε-acetyllysine residues on nucleosomal histones. Upon deacetylation of histone proteins, the interaction between histone proteins and DNA increases, which reduces DNA expression and gene transcription.1,2 In addition, HDAC proteins affect intracellular interactions, protein localization, and protein stability through deacetylation of non-histone substrates.36 Beyond basic cell biology, HDAC proteins are overexpressed in several cancers, as well as other diseases.7

HDAC proteins require either metal ions or NAD+ as cofactors for catalysis.8 The eleven metal-dependent HDAC isoforms are grouped as classes I, II, or IV, depending on their size, cellular localization, and homology to other HDAC proteins.9 The seven NAD+ dependent HDAC proteins are grouped as class III. The metal-dependent HDAC proteins comprise eleven isoforms (HDAC 1–11) and are the focus of this work.

Each metal-dependent HDAC isoform has been associated with various cancers. For example, HDAC1 was overexpressed in lung and colon cancers,10, 11 while HDAC2 displayed aberrant expression in colorectal and gastric cancer.12 Abnormal activity of HDAC8 was observed in acute myeloid leukemia, T-cell lymphoma, and neuroblastoma.13 HDAC6 was highly expressed in leukemia, ovarian cancer, and oral squamous cell carcinoma.1416 Moreover, HDAC6 plays an important role in cancer cell growth and survival through several non-epigenetic pathways.17 Importantly, HDAC6 and HDAC8 were expressed at abnormally high levels in various human breast cancer cell lines and were associated with breast cancer invasiveness and metastasis, making them both interesting for anti-cancer drug design.18

Due to their fundamental role in cancer, inhibitors targeting HDAC proteins have been developed. HDAC inhibitors showed the ability to reduce proliferation and metastasis, and promote apoptosis in several cancers.14, 1822 Four HDAC inhibitors have been approved by the FDA for cancer treatment.20, 2326 For example, SAHA (suberoylamide hydroxamic acid, Vorinostat, Zolinza™) and Belinostat (PXD101, Belodaq™)(Figure 1) were approved for treatment of T-cell lymphoma,2325 while Panobinostat (LBH-589, Farydak™)(Figure 1) was approved for treatment of multiple myloma.26 SAHA inhibits most of the eleven metal-dependent HDAC proteins, with only a modest selectivity against HDAC8.27, 28 The non-selectivity of SAHA might explain the side effects observed in clinic, but certainly limits the use of SAHA to study individual HDAC isoforms in cancer biology.29 In the last four years, several dual HDAC6/HDAC8 selective inhibitors have been developed.3033 Dual inhibition of HDAC6 and HDAC8 can possibly have synergistic therapeutic applications in treatment of various cancers, which can improve anti-cancer efficiency compared to current non-selective HDAC inhibitors.3034

Figure 1.

Figure 1

Chemical structures of the FDA-approved drugs SAHA, Belinostat, and Panobinistat, along with the C5-modified SAHA analogs reported here.

SAHA and most of the HDAC inhibitors have a similar pharmacophore that consists of three structural elements (Figure 1). The capping group interacts with solvent-exposed residues of the HDAC active site, while the metal binding group forms key interactions with the catalytic metal deeply buried in the active site (Figure 1). The linker region that connects the capping and the metal binding groups is positioned in the narrow hydrophobic active site channel. Both the capping and the metal binding groups have been modified extensively in HDAC inhibitor design.3540 In contrast, few studies report modification of the linker region.4144 To study the effect of substitution on the linker region, SAHA analogs substituted at carbon 2 (C2), 3 (C3), or 6 (C6) of the linker region were synthesized and screened (Figure 1). C2-hexyl SAHA displayed dual HDAC6/8 selectivity,44 C3-ethyl SAHA showed HDAC6 selectivity,42 and C6-butyl SAHA inhibited HDAC1 and 6 over HDAC3.43 The conclusion of this prior work is that modification of the SAHA linker can alter inhibitor specificity.

Guided by this prior work, here we explored the effect of substituents on the C5 position of SAHA (Figure 1). SAHA analogs substituted at the C5 were synthesized and tested for potency and selectivity both in vitro and in cellulo. Several analogs showed dual HDAC6/8 selectivity over HDAC1, 2, and 3, with a modest reduction in HDAC6 inhibition but enhanced HDAC8 inhibition compared to SAHA. This study documents that modifying the linker region of SAHA can alter its selectivity with minimal effect on potency.

Synthesis of C5-modified SAHA analogs

C5-modified SAHA analogs 1a–e were synthesized as shown in Scheme 1. The synthesis started from a coupling reaction of 4-pentenoic acid 2 with aniline using TBTU to obtain amide 3, which was then reacted with crotonaldehyde 4 via a cross metathesis reaction using second generation Grubbs' catalyst to afford the α,β-unsaturated aldehyde 5. Aldehyde 5 was substituted with different groups through a 1,4-conjugate addition using organolithium or organomagnesium cuprates to yield intermediates 6a–e. Horner–Wadsworth–Emmons olefination of 6a–e with trimethyl phosphonoacetate followed by reduction gave amide esters 8a–e with a saturated linker. Finally, amide esters 8a–e were reacted with hydroxylamine to afford the C5-substituted SAHA derivatives 1a–e as racemic mixtures.

Scheme 1.

Scheme 1

Synthesis of C5-modified SAHA analogs (1ae)

In vitro screening of C5-modified SAHA analogs

As a preliminary screen, the new analogs were tested for their global HDAC inhibition with HeLa cell lysates as the source of all HDAC proteins (Table 1). SAHA was also tested as the parent unsubstituted control molecule. The inhibitory activities of the analogs were measured with the HDAC-Glo™ I/II substrate (Promega). C5-methyl SAHA analog 1a showed greater potency compared to SAHA (100 nM vs 200 nM IC50 values, Table 1). However, all other analogs showed weaker potency than SAHA (11- to 33-fold reduction in potency), with IC50 values from 2.2 to 6.5 µM (Table 1). The observed lower potencies of compounds 1b1e may be due to selectivity for specific HDAC isoform(s), which lowered the potency against lysates that contains all HDAC isoforms. The lower potency observed here was similar to what was observed with the C2-modified SAHA analogs.44

Table 1.

IC50 values for SAHA and C5-modified SAHA analogs (1a1e) with HeLa cell lysates.a

Compounds R IC50 (µM)
SAHA H 0.20 ± 0.02
1a methyl 0.10 ± 0.01
1b n-butyl 5.0 ± 0.4
1c n-hexyl 6.5 ± 0.1
1d phenyl 2.2 ± 0.1
a

Mean IC50 value and standard error of at least three independent trials are shown (Figure S52 and Table S1).

To test isoform selectivity, the parent molecule, SAHA, and all the C5-modified SAHA analogs were tested at a single concentration using the recently developed ELISA-based HDAC activity assay (Figure 2).28 SAHA, as expected, showed no selectivity among HDAC1, 2, 3, and 6.28 Interestingly, several C5-SAHA analogs displayed more potent inhibition against HDAC6 compared to HDAC1, HDAC2, and HDAC3 (Figure 2). The analogs that showed the greater difference in potency with HDAC6 versus the other isoforms were C5-n-butyl (1b), C5-n-hexyl (1c), and C5-benzyl (1e). The C5-methyl SAHA (1a) and C5-phenyl SAHA (1d) showed only a small difference in potency comparing HDAC6 to the others (Figure 2).

Figure 2.

Figure 2

In vitro isoform selectivity screening of C5-modified SAHA analogs (1ae) against HDAC1, HDAC2, HDAC3, and HDAC6 using the ELISA-based HDAC activity assay. Analogs 1a–e were tested at 0.025, 0.25, 1.25, 0.125, and 1.25 µM final concentrations, respectively. SAHA was tested at 1 µM concentration in a previous report using the same assay procedure.28 Mean percent deacetylase activities from a minimum of two independent trials with standard errors were plotted (Table S2).

IC50 values for the most selective derivatives 1b, 1c, and 1e were determined with HDAC1, HDAC2, HDAC3, HDAC6, and HDAC8 isoforms to quantitatively assess the selectivity (Table 2). HDAC8 was also tested due to its similar active site compared to HDAC6. The IC50 values of SAHA as the parent compound were included as well (Table 2).28 SAHA displayed similar IC50 values against HDAC1, 2, 3, and 6, with 6- to 27-fold selectivity against HDAC8.28, 44 Both C5-n-butyl (1b), and C5-n-hexyl (1c) SAHA analogs displayed modest selectivity, with 3- to 5-fold and 5- to 7-fold selectivities for HDAC6 and HDAC8 over HDAC1, 2, and 3 (Tables 2 and S6). In addition, 1b and 1c showed modest reductions in HDAC6 potency (IC50 values of 320 and 410 nM), but similar potency against HDAC8 (430 and 420 nM) compared to SAHA (Table 2). The most potent and selective analog was C5-benzyl SAHA (1e), which displayed 8- to 21-fold selectivity for HDAC6 and HDAC8 over HDAC1, 2, and 3 and IC50 values of 270 and 380 nM with HDAC6 and HDAC8, respectively (Table 2 and S6). The selectivity was due to a dramatic reduction in potency with HDAC1, 2, and 3 (14- to 80-fold), but only a modest potency reduction with HDAC6 (8.5- to 12-fold) and similar potency with HDAC8 (380 – 540 nM), compared to SAHA. Modification of SAHA at the C5 position of the linker led to selectivity for HDAC6 and 8 over HDAC1, 2, and 3.

Table 2.

IC50 values for SAHA and C5-modified SAHA analogs 1b, 1c, and 1e against HDAC1, 2, 3, 6 and 8.a

Compound IC50 values (nM)

HDAC1 HDAC2 HDAC3 HDAC6 HDAC8

SAHAb 33 ± 1 96 ± 10 20 ± 1 33 ± 3 540 ± 10
1b (n-butyl) 1,100 ± 100 1,300 ± 100 1,600 ± 100 320 ± 30 430 ± 10
1c (n-hexyl) 2,100 ± 100 2,500 ± 100 2,900 ± 300 410 ± 60 420 ± 20
1e (benzyl) 2,900 ± 300 3,500 ± 100 5,800 ± 800 270 ± 20 380 ± 20
a

Mean IC50 value and standard error of at least three independent trials are shown (Figures S53–S55 and Tables S3–S5).

b

Previously reported IC50 values using the same assay procedure.28

The size of the substituent had a substantial effect on selectivity (Figure 2 and Table 2). More bulky or longer alkyl chain enhanced the selectivity to HDAC6 and 8 over HDAC1, 2, and 3. As suggested by previous reports,45 larger substituents in the linker region might have led to enhanced HDAC6/8 selectivity due to the wider active site entrance of both HDAC6 and HDAC8 compared to HDAC1, 2 and 3. Steric clash with the active sites of HDAC1, 2, and 3 might explain the reduction in inhibition of HDAC1, 2, and 3 (36- to 290-fold reduction in IC50 values, see Table 2).44, 46 Consistent with this analysis, docking studies of the structurally similar C2-hexyl SAHA analog showed that both enantiomers had a similar selectivity and IC50 values, which suggests that the size of the substituent is the origin of the observed selectivity rather than the chirality of the substituted carbon.44

In cellulo selectivity testing

To test the analogs in a more biological context, the C5-benzyl (1e) SAHA analog was tested for selectivity in cells. The inhibition of HDAC6 was monitored by detecting the levels of its known substrate acetyl-α-tubulin (AcTub), whereas Class I HDAC (HDAC1, 2, and 3) inhibition was monitored by observing the known substrate, acetyl-histone 3 (AcH3). SAHA or C5-benzyl SAHA 1e were incubated with U937 leukemia cells before lysis and western blot analysis of protein acetylation (Figure 3). As expected, SAHA increased the levels of both acetyl-α-tubulin and acetyl-histone H3 to a similar extent (Figure 3, lane 1), which is consistent with its nonselective inhibition of HDAC1, 2, and 3 isoforms. On the other hand, C5-benzyl SAHA analog 1e showed a dose dependent selective increase in levels of acetyl-α-tubulin, which was greater than the increased levels of acetyl histone H3 (Figure 3, lanes 3–5) compared to the DMSO control (Figure 3, lane 2). The observed HDAC6 selectivity of the C5-benzyl SAHA 1e in cells is consistent with the selectivity observed in the in vitro screening (Table 2 and Figure 3).

Figure 3.

Figure 3

Western blots analysis of acetyl-lysine 9 of histone H3, (AcH3) and acetyl-lysine 40 of α-tubulin (AcTub) after treatment with SAHA or C5-benzyl SAHA 1e. U937 cells were treated with DMSO (1%), SAHA (5 µM), or increasing concentrations of C5-benzyl SAHA (1e) analog (10–40 µM), before lysis, SDS-PAGE separation, transfer to a PVDF membrane, and western analysis with AcH3 or AcTub antibodies. GAPDH levels in the samples were also probed as a gel load control. A DMSO control sample was included for comparison to inhibitor-treated samples. Repetitive trials are shown in Figures S56.

In vitro cancer cell growth inhibition

To evaluate the ability of the C5-modified SAHA analogs to influence cell growth, the most selective analogs were tested. C5-n-butyl (1b), C5-n-hexyl (1c), and C5-benzyl (1e) SAHA analogs were tested at 1 and 10 µM concentrations using MTT assay. Jurkat cells, a T-cell lymphoma derived cancer cell line, were selected due to the role of both HDAC6 and HDAC8 in lymphoma.22, 47 The tested analogs displayed cytotoxicity against the cell line, with 39%, 24%, and 50% cell viability with C5-n-butyl SAHA (1b), C5-n-hexyl SAHA (1c), and C5-benzyl SAHA (1e) at 10 µM concentrations, respectively (Figure 4). On the other hand, SAHA demonstrated higher cytotoxicity than the C5-SAHA analogs, with 49% cell viability at 1 µM. The reduced cytotoxicity of the analogs compared to SAHA can be due to their lower potency (8- to 12-fold reduction in IC50 values with HDAC6 compared to SAHA, Table 2). In addition, the nonselective inhibition of most HDAC isoforms by SAHA might contribute to its higher cytotoxicity (Table 2 and Figure 4).

Figure 4.

Figure 4

Cytotoxicity screening of SAHA and C5-modified SAHA analogs 1b, 1c, and 1e, at 1 and 10 µM concentrations using an MTT assay with the Jurkat cells. Mean percent cell viability from a minimum of three independent trials with standard errors were plotted (Table S7).

In conclusion, C5-modified SAHA analogs displayed dual HDAC6/8 selectivity. The best compound was C5-benzyl SAHA (1e), which showed up to 21-fold selectivity for HDAC6 and 8 compared to HDAC1, 2, and 3, and IC50 values of 270 and 380 nM with HDAC6 and 8, respectively. C5-benzyl SAHA (1e) was tested for isoform selectivity in cells and showed selectivity consistent to what was observed in vitro. The fold selectivities observed with the C5-modified SAHA analogs were reduced compared to previously reported HDAC6/8-selective C2-modified SAHA analogs (49- to 300- fold selective for HDAC6 and 8 over HDAC1, 2, and 3 for the C2-hexyl SAHA).44 On the other hand, the C5-modified SAHA analogs were more potent against HDAC6 and 8 (270 to 430 nM IC50 values) compared to C2-modifed SAHA analogs (600 to 2,000 nM IC50 values for C2-hexyl SAHA). In general, this study and previous reports indicate that modifying SAHA in the linker region can alter the selectivity of HDAC inhibitors. In particular, the reduced potency of SAHA against HDAC8 compared to HDAC1, 2, and 3 was switched in the C5-modified analogs; C5-benzyl SAHA (1e) showed dramatically reduced potency for HDAC1, 2, and 3, but similar HDAC8 and modestly lower HDAC6 potency, compared to SAHA, which resulted in dual HDAC6/8 preference. HDAC6/8 dual selective inhibitors can be used as biological tools to study both HDAC6 and HDAC8-related cancer biology, and as leads for development of more effective anti-cancer agents targeting both HDAC6 and HDAC8.

Supplementary Material

supplement

Acknowledgments

Funding Sources

We thank the National Institutes of Health (GM121061) and Wayne State University for funding. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

We thank Lumigen Instrument Center at Wayne State University for NMR and MS instrumentation, Dr. K. Honn and Dr. Y. Ahn for use of the Alpha Innotech FluorChem imaging systems, and I. Gomes, J. Knoff, D. Nalawansha, and Y. Zhang for comments on the manuscript.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Supplementary Material

Supplementary material (experimental procedures, compound synthesis and characterization, procedure and data for biological screening, and IC50 curves and tables) associated with this article can be found with the online version.

References

  • 1.Walsh CT, Garneau-Tsodikova S, Gatto GJ. Protein posttranslational modifications: the chemistry of proteome diversifications. Angew Chem, Int Ed. 2005;44(45):7342–7372. doi: 10.1002/anie.200501023. [DOI] [PubMed] [Google Scholar]
  • 2.Hebbes TR, Thorne AW, Crane-Robinson C. A direct link between core histone acetylation and transcriptionally active chromatin. EMBO J. 1988;7(5):1395–1402. doi: 10.1002/j.1460-2075.1988.tb02956.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Nalawansha DA, Pflum MKH. LSD1 Substrate Binding and Gene Expression Are Affected by HDAC1-Mediated Deacetylation. ACS chemical biology. 2017;12(1):254–264. doi: 10.1021/acschembio.6b00776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Chuang C, Pan J, Hawke DH, Lin S-H, Yu-Lee L-y. NudC deacetylation regulates mitotic progression. PLoS ONE. 2013;8(9):e73841. doi: 10.1371/journal.pone.0073841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Martinez-Balbas MA, Bauer UM, Nielsen SJ, Brehm A, Kouzarides T. Regulation of E2F1 activity by acetylation. The EMBO journal. 2000;19(4):662–671. doi: 10.1093/emboj/19.4.662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ito A, Kawaguchi Y, Lai CH, et al. MDM2–HDAC1-mediated deacetylation of p53 is required for its degradation. The EMBO journal. 2002;21(22):6236–6245. doi: 10.1093/emboj/cdf616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Glozak MA, Seto E. Histone deacetylases and cancer. Oncogene. 2007;26(37):5420–5432. doi: 10.1038/sj.onc.1210610. [DOI] [PubMed] [Google Scholar]
  • 8.Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M. Epigenetic protein families: a new frontier for drug discovery. Nat Rev Drug Discov. 2012;11(5):384–400. doi: 10.1038/nrd3674. [DOI] [PubMed] [Google Scholar]
  • 9.Gregoretti I, Lee Y-M, Goodson HV. Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis. J Mol Biol. 2004;338(1):17–31. doi: 10.1016/j.jmb.2004.02.006. [DOI] [PubMed] [Google Scholar]
  • 10.Bartling B, Hofmann HS, Boettger T, et al. Comparative application of antibody and gene array for expression profiling in human squamous cell lung carcinoma. Lung Cancer. 2005;49(2):145–154. doi: 10.1016/j.lungcan.2005.02.006. [DOI] [PubMed] [Google Scholar]
  • 11.Wilson AJ, Byun D-S, Popova N, et al. Histone deacetylase 3 (HDAC3) and other Class I HDACs regulate colon cell maturation and p21 expression and are deregulated in human colon cancer. J Biol Chem. 2006;281(19):13548–13558. doi: 10.1074/jbc.M510023200. [DOI] [PubMed] [Google Scholar]
  • 12.Huang BH, Laban M, Leung CH-W, et al. Inhibition of histone deacetylase 2 increases apoptosis and p21Cip1/WAF1 expression, independent of histone deacetylase 1. Cell Death Differ. 2005;12(4):395–404. doi: 10.1038/sj.cdd.4401567. [DOI] [PubMed] [Google Scholar]
  • 13.Oehme I, Deubzer HE, Wegener D, et al. Histone deacetylase 8 in neuroblastoma tumorigenesis. Clin Cancer Res. 2009;15(1):91–99. doi: 10.1158/1078-0432.CCR-08-0684. [DOI] [PubMed] [Google Scholar]
  • 14.Hackanson B, Rimmele L, Benkißer M, et al. HDAC6 as a target for antileukemic drugs in acute myeloid leukemia. Leuk Res. 2012;36(8):1055–1062. doi: 10.1016/j.leukres.2012.02.026. [DOI] [PubMed] [Google Scholar]
  • 15.Sakuma T, Uzawa K, Onda T, et al. Aberrant expression of histone deacetylase 6 in oral squamous cell carcinoma. Int J Oncol. 2006;29(1):117–124. [PubMed] [Google Scholar]
  • 16.Bazzaro M, Lin Z, Santillan A, et al. Ubiquitin Proteasome System Stress Underlies Synergistic Killing of Ovarian Cancer Cells by Bortezomib and a Novel HDAC6 Inhibitor. Clinical Cancer Research. 2008;14(22):7340–7347. doi: 10.1158/1078-0432.CCR-08-0642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Aldana-Masangkay GI, Sakamoto KM. The role of HDAC6 in cancer. J Biomed Biotechnol. 2011;2011:875824. doi: 10.1155/2011/875824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Park SY, Jun JA, Jeong KJ, et al. Histone deacetylases 1, 6 and 8 are critical for invasion in breast cancer. Oncol Rep. 2011;25(6):1677–1681. doi: 10.3892/or.2011.1236. [DOI] [PubMed] [Google Scholar]
  • 19.Johnstone RW. Histone-deacetylase inhibitors: novel drugs for the treatment of cancer. Nat Rev Drug Discov. 2002;1(4):287–299. doi: 10.1038/nrd772. [DOI] [PubMed] [Google Scholar]
  • 20.Rodd AL, Ververis K, Karagiannis TC. Current and emerging therapeutics for cutaneous T-Cell lymphoma: histone deacetylase inhibitors. Lymphoma. 2012;2012:10. [Google Scholar]
  • 21.West AC, Johnstone RW. New and emerging HDAC inhibitors for cancer treatment. J Clin Invest. 2014;124(1):30–39. doi: 10.1172/JCI69738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Balasubramanian S, Ramos J, Luo W, Sirisawad M, Verner E, Buggy JJ. A novel histone deacetylase 8 (HDAC8)-specific inhibitor PCI-34051 induces apoptosis in T-cell lymphomas. Leukemia. 2008;22:1026–1034. doi: 10.1038/leu.2008.9. [DOI] [PubMed] [Google Scholar]
  • 23.Grant S, Easley C, Kirkpatrick P. Vorinostat. Nat Rev Drug Discovery. 2007;6(1):21–22. doi: 10.1038/nrd2227. [DOI] [PubMed] [Google Scholar]
  • 24.Plumb JA, Finn PW, Williams RJ, et al. Pharmacodynamic response and inhibition of growth of human tumor xenografts by the novel histone deacetylase inhibitor PXD101. Mol Cancer Ther. 2003;2(8):721–728. [PubMed] [Google Scholar]
  • 25.Prince HM, Dickinson M. Romidepsin for cutaneous T-cell lymphoma. Clinical cancer research : an official journal of the American Association for Cancer Research. 2012;18(13):3509–3515. doi: 10.1158/1078-0432.CCR-11-3144. [DOI] [PubMed] [Google Scholar]
  • 26.Laubach JP, Moreau P, San-Miguel JF, Richardson PG. Panobinostat for the treatment of multiple myeloma. Clin Cancer Res. 2015;21(21):4767–4773. doi: 10.1158/1078-0432.CCR-15-0530. [DOI] [PubMed] [Google Scholar]
  • 27.Khan N, Jeffers M, Kumar S, et al. Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors. Biochem J. 2008;409(2):581–589. doi: 10.1042/BJ20070779. [DOI] [PubMed] [Google Scholar]
  • 28.Padige G, Negmeldin AT, Pflum MKH. Development of an ELISA-based HDAC activity assay for characterization of isoform-selective inhibitors. J Biomol Screening. 2015;20(10):1277–1285. doi: 10.1177/1087057115598118. [DOI] [PubMed] [Google Scholar]
  • 29.O'Connor OA, Heaney ML, Schwartz L, et al. Clinical experience with intravenous and oral formulations of the novel histone deacetylase inhibitor suberoylanilide hydroxamic acid in patients with advanced hematologic malignancies. Journal of Clinical Oncology. 2006;24(1):166–173. doi: 10.1200/JCO.2005.01.9679. [DOI] [PubMed] [Google Scholar]
  • 30.Olson DE, Wagner FF, Kaya T, et al. Discovery of the first histone deacetylase 6/8 dual inhibitors. J Med Chem. 2013;56(11):4816–4820. doi: 10.1021/jm400390r. [DOI] [PubMed] [Google Scholar]
  • 31.Tang G, Wong JC, Zhang W, et al. Identification of a Novel Aminotetralin Class of HDAC6 and HDAC8 Selective Inhibitors. Journal of medicinal chemistry. 2014;57(19):8026–8034. doi: 10.1021/jm5008962. [DOI] [PubMed] [Google Scholar]
  • 32.Rodrigues DA, Ferreira-Silva GÀ, Ferreira ACS, et al. Design, Synthesis, and Pharmacological Evaluation of Novel N-Acylhydrazone Derivatives as Potent Histone Deacetylase 6/8 Dual Inhibitors. Journal of medicinal chemistry. 2016;59(2):655–670. doi: 10.1021/acs.jmedchem.5b01525. [DOI] [PubMed] [Google Scholar]
  • 33.Suzuki T, Ota Y, Ri M, et al. Rapid Discovery of Highly Potent and Selective Inhibitors of Histone Deacetylase 8 Using Click Chemistry to Generate Candidate Libraries. Journal of medicinal chemistry. 2012;55(22):9562–9575. doi: 10.1021/jm300837y. [DOI] [PubMed] [Google Scholar]
  • 34.Oehme I, Deubzer HE, Lodrini M, Milde T, Witt O. Targeting of HDAC8 and investigational inhibitors in neuroblastoma. Expert Opin Invest Drugs. 2009;18(11):1605–1617. doi: 10.1517/14728220903241658. [DOI] [PubMed] [Google Scholar]
  • 35.Estiu G, Greenberg E, Harrison CB, et al. Structural origin of selectivity in class II-selective histone deacetylase inhibitors. J Med Chem. 2008;51(10):2898–2906. doi: 10.1021/jm7015254. [DOI] [PubMed] [Google Scholar]
  • 36.Bieliauskas AV, Weerasinghe SVW, Negmeldin AT, Pflum MKH. Structural requirements of histone deacetylase inhibitors: SAHA analogs modified on the hydroxamic acid. Arch Pharm (Weinheim, Ger) 2016;349(5):373–382. doi: 10.1002/ardp.201500472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Suzuki T, Kouketsu A, Matsuura A, et al. Thiol-based SAHA analogues as potent histone deacetylase inhibitors. Bioorganic & Medicinal Chemistry Letters. 2004;14(12):3313–3317. doi: 10.1016/j.bmcl.2004.03.063. [DOI] [PubMed] [Google Scholar]
  • 38.Kapustin GV, Fejér G, Gronlund JL, McCafferty DG, Seto E, Etzkorn FA. Phosphorus-Based SAHA Analogues as Histone Deacetylase Inhibitors. Organic Letters. 2003;5(17):3053–3056. doi: 10.1021/ol035056n. [DOI] [PubMed] [Google Scholar]
  • 39.Salmi-Smail C, Fabre A, Dequiedt F, et al. Modified Cap Group Suberoylanilide Hydroxamic Acid Histone Deacetylase Inhibitor Derivatives Reveal Improved Selective Antileukemic Activity. Journal of medicinal chemistry. 2010;53(8):3038–3047. doi: 10.1021/jm901358y. [DOI] [PubMed] [Google Scholar]
  • 40.Oger F, Lecorgne A, Sala E, et al. Biological and Biophysical Properties of the Histone Deacetylase Inhibitor Suberoylanilide Hydroxamic Acid Are Affected by the Presence of Short Alkyl Groups on the Phenyl Ring. Journal of medicinal chemistry. 2010;53(5):1937–1950. doi: 10.1021/jm901561u. [DOI] [PubMed] [Google Scholar]
  • 41.Bieliauskas A, Weerasinghe S, Pflum MH. Structural requirements of HDAC inhibitors: SAHA analogs functionalized adjacent to the hydroxamic acid. Bioorg Med Chem Lett. 2007;17(8):2216–2219. doi: 10.1016/j.bmcl.2007.01.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Choi SE, Weerasinghe SV, Pflum MK. The structural requirements of histone deacetylase inhibitors: Suberoylanilide hydroxamic acid analogs modified at the C3 position display isoform selectivity. Bioorg Med Chem Lett. 2011;21(20):6139–6142. doi: 10.1016/j.bmcl.2011.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Choi SE, Pflum MKH. The structural requirements of histone deacetylase inhibitors: Suberoylanilide hydroxamic acid analogs modified at the C6 position. Bioorg Med Chem Lett. 2012;22(23):7084–7086. doi: 10.1016/j.bmcl.2012.09.093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Negmeldin AT, Padige G, Bieliauskas AV, Pflum MKH. Structural Requirements of HDAC Inhibitors: SAHA Analogues Modified at the C2 Position Display HDAC6/8 Selectivity. ACS Medicinal Chemistry Letters. 2017;8(3):281–286. doi: 10.1021/acsmedchemlett.6b00124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Negmeldin AT, Padige G, Bieliauskas AV, Pflum MKH. Structural Requirements of HDAC Inhibitors: SAHA Analogues Modified at the C2 Position Display HDAC6/8 Selectivity. ACS Medicinal Chemistry Letters. 2017 doi: 10.1021/acsmedchemlett.6b00124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Fass DM, Shah R, Ghosh B, et al. Short-chain HDAC inhibitors differentially affect vertebrate development and neuronal chromatin. ACS Med Chem Lett. 2011;2(1):39–42. doi: 10.1021/ml1001954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Amengual JE, Johannet P, Lombardo M, et al. Dual Targeting of Protein Degradation Pathways with the Selective HDAC6 Inhibitor ACY-1215 and Bortezomib Is Synergistic in Lymphoma. Clinical Cancer Research. 2015;21(20):4663–4675. doi: 10.1158/1078-0432.CCR-14-3068. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

supplement

RESOURCES